You are on page 1of 12

International Journal of

Molecular Sciences

Article
Validation Study of a New Random-Access Chemiluminescence
Immunoassay Analyzer i-TRACK10® to Monitor Infliximab and
Adalimumab Serum trough Levels and Anti-Drug Antibodies
Anne Emmanuelle Berger 1,2,3 , Aude Gleizes 4,5,6 , Louis Waeckel 1,2,3 , Xavier Roblin 1,2,7 , Roman Krzysiek 4,8 ,
Salima Hacein-Bey-Abina 4,5 , Alessandra Soriano 9, * and Stephane Paul 1,2,3

1 CIRI—Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Université Claude


Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, F69007 Lyon, France
2 CIC Inserm 1408 Vaccinology, F42023 Saint-Etienne, France
3 Department of Immunology, iBIOTHERA Reference Center, University Hospital of Saint-Etienne,
F42023 Saint-Etienne, France
4 Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Saclay, Hôpital Bicêtre, Assistance
Publique-Hôpitaux de Paris, F94270 Paris, France
5 Unité des Technologies Chimiques et Biologiques pour la Santé, Université de Paris, CNRS, INSERM, UTCBS,
F75270 Paris, France
6 Faculty of Pharmacy, Paris-Saclay University, F92290 Paris, France
7 Gastroenterology Department, CHU Saint-Etienne, F42023 Saint-Etienne, France
8 Faculté de Médecine, Université Paris-Saclay, UMR-996 INSERM Inflammation, Microbiome and
Immunosurveillance, F92141 Clamart, France
9 Gastroenterology Division and IBD Center, Internal Medicine Department, Azienda Unità Sanitaria
Citation: Berger, A.E.; Gleizes, A.; Locale—IRCCS, 42122 Reggio Emilia, Italy
Waeckel, L.; Roblin, X.; Krzysiek, R.; * Correspondence: alessandra.soriano@ausl.re.it
Hacein-Bey-Abina, S.; Soriano, A.;
Paul, S. Validation Study of a New Abstract: Background. Monitoring of biological TNF inhibitors is a very important tool to guide clin-
Random-Access Chemiluminescence ical decisions using specialized algorithms, especially in gastroenterology. A new chemiluminescent
Immunoassay Analyzer i-TRACK10® instrument (i-TRACK10® from Theradiag) could replace ELISA techniques to calculate the dosage
to Monitor Infliximab and of drugs and anti-drug antibodies. In this bi-centric study, we explored the analytical performances
Adalimumab Serum trough Levels
of i-TRACK10® using manual or automated (DS2® ) ELISA Lisa-Tracker® assays, and compared the
and Anti-Drug Antibodies. Int. J. Mol.
results. Patients and methods. Intra- and inter-run performances were evaluated with i-TRACK10®
Sci. 2022, 23, 9561. https://doi.org/
in two different laboratories and for two different ranges of values for infliximab, adalimumab,
10.3390/ijms23179561
and their respective antibodies. Patients’ samples were used in the labs to compare the results
Academic Editor: Maria obtained between the new instrument and either the manual Lisa-Tracker® or the automated DS2.
Grazia Cifone Results. Intra- and inter-run performances were satisfactory, with values between 1.8% and 16.1%
Received: 19 July 2022 (for inter-run imprecision at low/medium values of infliximab). Results were generally comparable
Accepted: 20 August 2022 between assays. with the lowest value of correlation at 0.59 (anti-adalimumab dosage between
Published: 24 August 2022 i-TRACK10® and manual ELISA). Most often, values of drugs and anti-drug antibodies were higher
with i-TRACK10® than with manual ELISA assay, and correlation values were better with automated
Publisher’s Note: MDPI stays neutral
ELISA. Agreements were globally acceptable, and the lowest coefficients of 0.7 was obtained for adal-
with regard to jurisdictional claims in
published maps and institutional affil-
imumab values between i-TRACK10® and the two ELISA methods, and for anti-adalimumab values
iations. between i-TRACK10® and manual ELISA. The type of assay can potentially induce a change in the
class of patients and lead to divergent therapeutic decisions. Conclusions. The new random-access
instrument i-TRACK10® presents many advantages in a routine laboratory: rapidity, the possibility
of standardization, usability, and expansion of the measurement range. Despite the relatively good
Copyright: © 2022 by the authors. agreement of results, it is preferable to use the same assay in longitudinal follow-up of a patient,
Licensee MDPI, Basel, Switzerland. because quantitative results were not completely equivalent especially for anti-drug antibodies.
This article is an open access article
distributed under the terms and
Keywords: TNF inhibitors; adalimumab; infliximab; therapeutic drug monitoring; anti-drug antibodies;
conditions of the Creative Commons
CLIA; ELISA
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).

Int. J. Mol. Sci. 2022, 23, 9561. https://doi.org/10.3390/ijms23179561 https://www.mdpi.com/journal/ijms


Int. J. Mol. Sci. 2022, 23, 9561 2 of 12

1. Background
Tumor necrosis factor (TNF) is recognized as a key player in a broad range of immune
inflammatory diseases. The advent of TNF inhibitors (TNFi) has progressively changed the
therapeutic field, notably for inflammatory bowel and rheumatologic diseases [1]. Anti-TNF
biologics have proved to be very effective in the induction and maintenance of remission in
patients who do not respond to conventional immunosuppressive therapy [2]. Presently,
there are five TNFi approved by EMA and FDA: adalimumab (ADAL); golimumab (another
fully human IgG1 monoclonal antibody); infliximab (IFX) (a chimeric IgG1 monoclonal
antibody); certolizumab (a PEGylated Fab fragment of a humanized anti-TNF monoclonal
antibody); and etanercept (a fusion protein between a human IgG1 Fc-tail and human
TNF-receptor type 2/TNFR2).
Although TNFis have significantly improved treatment outcomes for chronic inflam-
matory diseases, some patients have had to discontinue treatment due to primary non-
response, secondary treatment failure, or adverse reactions. Primary non-response can
occur in about 1/3 of patients, and up to 60% of initial responders lose response over time,
defined as a need for dose escalation and drug discontinuation rates. A high suboptimal
response rate or lack of response to first-line therapy with TNFi can be caused by non-
immune related factors that affect drug clearance, such as high body mass index (BMI) or
high disease burden. However, one of the main reasons for the loss of therapeutic efficacy
of TNFis (secondary failure) is immunogenicity, namely the development of anti-drug
antibodies (ADAs), especially drug-neutralizing ADAs against chimeric, humanized, or
fully human TNFi [3].
ADAs have been linked to subtherapeutic serum drug levels by increasing the clear-
ance and reducing the bioavailability of the TNFi, thus determining loss of clinical response.
Moreover, ADAs have been linked to infusion-related adverse events, including hypersen-
sitivity reactions [4]. Close monitoring of patients’ TNFi trough levels (i.e., therapeutic drug
monitoring or TDM) has been proven to be a valuable tool to aid therapy optimization, as
well as being a cost-effective strategy [5,6].
To address loss of response, several TDM algorithms have been proposed, which
commonly involve assessment of drug trough concentrations and ADA levels. Changing
the dosing regimen or dose intensity is often the first choice for treatment optimization.
Other options, such as adding immunosuppressive co-medication, switching to a different
TNFi (cycling strategy), or different classes of immunomodulatory drugs, can be potentially
considered, especially in patients who develop ADAs.
Reliable assays measuring TNFi with accuracy and precision are a prerequisite for
successful TDM. Several methods have been described to measure ADA levels, including
different formats of ELISA, radioimmunoassays, liquid-chromatography-based homoge-
neous mobility shift assay (HMSA), or chemiluminescence immunoassays (CLIA). These
methods use different designs and detection antibodies, and are differently prone to inter-
ference by TNFi, rheumatoid factors (RFs), or other interference factors [7]. Currently in
clinical practice, ELISA tests represent the most commonly used type of assay for TDM
in TNFi-treated patients. However, these assays often require several hours to complete
without random access flexibility functioning. This may limit the effectiveness of TDM by
significantly delaying dose adjustment in the subsequent drug administration, or delaying
changes in treatment strategy.
This study aimed to evaluate the performance of the latest generation chemilumines-
cence random-access immunoassay analyzer i-TRACK10® (Theradiag). Its performance
was compared with the currently used Lisa-Tracker® ELISA assays, which were performed
manually or automatically using the DS2 ELISA processing instrument (Theradiag). These
assays were used to monitor trough levels of IFX or ADAL, and ADA concentrations in
TNFi-treated patients.
Int. J. Mol. Sci. 2022, 23, 9561 3 of 12

2. Results
2.1. Sample and Assay Characteristics
The key features of the analytical systems used in the study are summarized in
Table 1. The main difference was the extended measurement range demonstrated by the
i-TRACK10® analyzer, both for serum drug concentration and ADA quantification. There
was no difference for the coated antigen used (human recombinant TNF for IFX and ADAL
assays, and the respective drugs for the aIFX and aADAL assays). No interference with
hemolysis, bilirubin, triglyceride, rheumatoid factors, or biotin in the sample matrix has
been reported for the assays analyzed in the study. Importantly, no significant cross-
reactivity has been described with other TNFis, other biological classes, or molecules
within the same disease spectrum. In the absence of prior dissociation of the immune
complexes formed by the drugs and ADAs, the assays were considered drug-sensitive
(reduced sensitivity to detect ADAs in the presence of the free drug).

Table 1. Assay characteristics.

Antigen Method Measurement Range Interference


aIFX/aADAL
IFX/ADAL IFX ADAL aIFX aADAL
ADAs
No influence
onhaemolysis,
Manual
Lisa Human bilirubin, triglyceride,
Infliximab or
Tracker® recombinant 0.3–20 µg/mL 10–200 ng/mL 10–160 ng/mL RF. No cross-reaction
Adalimumab automated
assays TNF with other anti-TNF
(DS2) ELISA
molecules nor with
rituximab.
No influence of
haemolysis
(2 mg/mL), bilirubin
(0.2 mg/mL),
triglyceride
(10 mg/mL), RF
(1000 UI/mL), biotin
Human
i-TRACK10® Infliximab (2000 ng/mL).
recombinant CLIA 0.3–24 µg/mL 0.5–24 µg/mL 10–2000 ng/mL
assay Adalimumab No cross-reactivity
TNF
with other anti-TNF
biologics, ustek-
inumab/vedolizumab,
nor with aADAL
ADAs (for IFX), nor
with aIFX ADAs (for
ADAL).
CLIA: ChemiLuminescence ImmunoAssay; ELISA: Enzyme-Linked ImmunoAssay; IFX: infliximab; ADAL:
adalimumab; aIFX: anti-infliximab antibodies; aADAL: anti-adalimumab antibodies; ADAs: anti-drug antibodies;
RF: rheumatoid factors. Source: Technical material for Lisa Tracker® and i-TRACK10® instruments.

2.2. Imprecision
The overall mean concentrations for each parameter (IFX, ADAL, and ADAs) and
the imprecision values are represented in Table 2 and Supplementary Figure S1. Intra-
and inter-run imprecisions were acceptable for all the assays (CV ranging from 1.8%
to 16.1%) according to the current FDA guidelines [8]. Indeed, 20% is defined as the
limit of acceptable CV% for precision and accuracy. Intra- and inter-run variations were
significantly different for all the assays, except IFX and aIFX measurement at the high level
solely for the B laboratory. The highest value was obtained for the inter-run imprecision of
IFX in laboratory B. Intra-run CV ranged from 1.8% to 11.3%, and the mean CV was 8.6%
for drugs and 2.7% for ADA quantification. Inter-run CV ranged from 7.3% to 16.1%, and
the mean CV was 11.2% for drugs and 7.9% for ADA quantification.
Int. J. Mol. Sci. 2022, 23, 9561 4 of 12

Table 2. Imprecisions of the i-TRACK10® instrument with the use of sample patients or QC
(low/medium and high values). For each analyte, the level of concentration used for the assay
and the obtained CVs are mentioned. ND: Not determinated.

Laboratory A Laboratory B
Low or Medium High Low or Medium High
Mean IFX—µg/mL
1.6 (8.2) 9.1 (11.3) 2.1 (5.8) 10.7 (8.3)
(Intra-run—CV%)
Mean IFX—µg/mL
2.3 (11.7) 11.7 (7.3) 2.3 (16.1) 10.3 (10)
(Inter-run—CV%)
Mean ADAL—µg/mL
1 (8.1) 13.3 (10.3) ND ND
(Intra-run—CV%)
Mean ADAL—µg/mL
3.7 (9.3) 12.5 (12.8)
(Inter-run—CV%)
Mean aIFX
ADAs—ng/mL 30 (3.7) 102 (3.7) 56 (1.8) 586 (2.6)
(Intra-run—CV%)
Mean aIFX
ADAs—ng/mL 60 (6.3) 603 (4.8) 61 (13.5) 594 (10.2)
(Inter-run—CV%)
Mean aADAL
ADAs—ng/mL 37 (1.8) 212 (2.2) ND ND
(Intra-run—CV%)
Mean aADAL
ADAs—ng/mL 53 (4.1) 505 (8.6)
(Inter-run—CV%)

2.3. Comparison of IFX and ADAL trough Levels


2.3.1. Comparison between i-TRACK10® and Lisa Tracker® for IFX and ADAL Quantification
The results for each assay were distributed into three categories: sub-therapeutic
(<3 µg/mL for IFX and <5 µg/mL for ADAL), optimal range (3–7 µg/mL for IFX and
5–8 µg/mL for ADAL), and high range (>7 µg/mL for IFX and >8 µg/mL for ADAL)
(Table 3). The kappa values were satisfactory according to the recommendations of Landis
et al. [9], and slightly better between i-TRACK10® and DS2® LT® (kappa value = 0.97) than
manual LT® (kappa value = 0.81) for IFX quantification. A change of classification for two
samples was observed between i-TRACK10® and manual LT® . Results were considered
in the high range for i-TRACK10® and in the optimal range for manual LT® . Change of
classification for one sample was observed between the i-TRACK10® and DS2 LT® results.
The sample was considered in the sub-therapeutic range by i-TRACK10® and in the optimal
range by DS2® LT® .
For ADAL quantification, the kappa value was slightly lower but was satisfactory and
similar to the two LT® methods (kappa value = 0.7). Comparing the results obtained with
i-TRACK10® and manual LT® , a change of classification was observed for three samples:
two that were in the optimal range according to i-TRACK10® and were found to be in the
sub-therapeutic range by the manual LT® assay, and one sample that was in the high range
with i-TRACK10® and in the optimal range with the manual LT® assay. The comparison
between i-TRACK10® and DS2® LT® showed ten particular discrepancies: six samples were
in the sub-therapeutic range with i-TRACK10® and in the optimal range with DS2® LT® ,
two samples that were in the high range level according to i-TRACK10® were found by
DS2® LT® to be in the optimal range, and two samples were in the optimal range with
i-TRACK10® and in the high range with DS2® LT® .
Int. J. Mol. Sci. 2022, 23, 9561 5 of 12

Table 3. Data agreement between i-TRACK10® and manual/DS2 Lisa Tracker® (LT) values for IFX,
ADAL, aIFX, and aADAL quantifications, according to therapeutic window stratification. The number
of samples that changed classification is indicated in bold and underlined.

IFX I-TRACK10
Manual LT
<3 µg/mL 3–7 µg/mL >7 µg/mL TOTAL
DS2 LT
2 0 0 2
<3 µg/mL
7 0 0 7
0 5 2 7
3–7 µg/mL
1 24 0 25
0 0 11 11
>7 µg/mL
0 0 18 18
2 5 13 20
TOTAL
8 24 18 50
0.81
Kappa value
0.97
ADAL I-TRACK10
Manual LT
<5 µg/mL 5–8 µg/mL >8 µg/mL TOTAL
DS2 LT
3 2 0 5
<5 µg/mL
7 0 0 7
0 1 1 2
5–8 µg/mL
6 8 2 16
0 0 13 13
>8 µg/mL
0 2 30 32
3 3 14 20
TOTAL
13 10 32 55
0.7
Kappa value
0.66
aIFX I-TRACK10
Manual LT
<100 ng/mL ≥100 ng/mL TOTAL
DS2 LT
10 0 10
<100 ng/mL
9 0 9
1 7 8
≥100 ng/mL
0 9 9
11 7 18
TOTAL
9 9 18
0.89
Kappa value
1
aADAL I-TRACK10
Manual LT
<100 ng/mL ≥100 ng/mL TOTAL
DS2 LT
35 4 39
<100 ng/mL
4 0 4
2 9 11
≥100 ng/mL
0 2 2
37 13 50
TOTAL
4 2 6
0.67
Kappa value
1

2.3.2. Correlation between i-TRACK10® and Lisa-Tracker® Assay Results


Linear regressions and Bland–Altman plots for each pair of assays are represented in
Figures 1a–d and 2a–d, respectively. Excellent linear correlations, between 0.85 and 0.92,
Int. J. Mol. Sci. 2022, 23, 9561 6 of 12

were obtained for each pair of assays. Globally, for all comparisons, the difference between
assays was slightly more prominent for the highest values of IFX and ADAL levels, but
remained acceptable. Analysis of systematic bias from the Bland–Altman plots indicated
that IFX levels were 17% higher on average in the i-TRACK10® group compared to data
obtained from the manual LT® (95% CI: −9.1%, 43.6%), with one point outside the 95% CI
(−0.9, 3; bias 1, Figure 2a). The results for the IFX performed with i-TRACK10® and DS2®
LT® were closer to similarity. Bland–Altman plot analysis showed that recorded IFX levels
were on average 10% lower using i-TRACK10® (95% CI: −46.7%, 26.5%), and the values
were aggregated, especially within the lower range (under 10 µg/mL). There were three
points outside the 95% CI (−3.7, 2.3; bias −0.7), which were connected to results within a
higher range of the values (Figure 2b). Average ADAL levels were 28% higher according to
i-TRACK10® compared with results obtained using manual LT® (95% CI: −2.2%, 58.1%),
with one point outside the 95% CI (−1.5, 7, bias 2.8, Figure 2c). The comparison of ADAL
levels between i-TRACK10® and DS2® LT® was similar, with the results of ADAL levels on
average 8% lower using i-TRACK10® (95% CI: −55.7%, 40.2%) compared with DS2® LT® ,
and with two points lying outside the 95% CI range (−4.1, 3.1; bias −0.5), Figure 2d.

Figure 1. (a–h) Comparison between IFX, ADAL, aIFX, and aADAL data obtained using i-TRACK10®
and Lisa-Tracker® (LT) assay performed manually or with the DS2 instrument. R2 values are shown
for all linear correlations (a–h).
Figure 1. (a–h) Comparison between IFX, ADAL, aIFX, and aADAL data obtained using i-TRACK10®
Int. J. Mol. Sci. 2022, 23, 9561 7 of 12
and Lisa-Tracker® (LT) assay performed manually or with the DS2 instrument. R²values are shown
for all linear correlations (a–h).

Figure 2. Bland–Altman plots to compare different assays: (a,b) Comparison of IFX level between
i-TRACK
i-TRACK10®10®and
andLisa
Lisatracker
tracker ® (LT)
® (LT) assays; (c,d)
assays; comparison
(c,d) of ADAL
comparison level
of ADAL between
level i-TRACK
between 10® and
i-TRACK 10®
Lisa-Tracker ®
and Lisa-Trackerassays;
® (e,f) comparison
assays; of aIFXof
(e,f) comparison level
aIFXbetween i-TRACKi-TRACK
level between
10® and LT assays;
®
10® and LT(g,h)
® com-
assays;
parison of aADALof
(g,h) comparison level
aADALbetween i-TRACK
level
10® and LT® assays. The difference between the two meas-
between i-TRACK 10® and LT® assays. The difference between
urements (µ g/mL for drugs and ng/mL for ADAs) is plotted on the y-axis, and the average of the
the two measurements (µg/mL for drugs and ng/mL for ADAs) is plotted on the y-axis, and the
two measurements on the x-axis. Dashed lines represent the bias and the 95% limits of agreement
average of the two measurements on the x-axis. Dashed lines represent the bias and the 95% limits of
for each comparison.
agreement for each comparison.

2.4. Comparison of Anti-IFX and Anti-ADAL ADA Testing


2.4.1. Good Agreement between i-TRACK10® and Lisa-Tracker® Measurements of aIFX
and aADAL
To render the results clinically relevant for patient-centered outcomes, a value of
100 ng/mL of ADAs was chosen to distinguish between low or moderate levels—which
could represent low affinity or transient ADAs, and high ADA values which would be
associated with a decrease of circulating TNFis and a loss of treatment efficacy. For the drug
assay, the agreement between i-TRACK10® and DS2® LT® for aIFX and aADAL (kappa
values = 1) was better than with the manual LT® assay (kappa values = 0.89 and 0.67
for aIFX and aADAL, respectively) (Table 3). Nevertheless, the results obtained with i-
TRACK10® agreed well with those obtained by the two LT assays, as previously reported
by Landis et al. [8]. Concerning aIFX, a change of classification for only one sample was
observed between i-TRACK10® and manual LT® . The result was considered within the
low/middle value range according to i-TRACK10® , and within the high range by manual
LT® . Concerning aADAL, a change of classification was observed for six samples: four
samples were considered within the low/middle range values by manual LT® and within
the higher range values by i-TRACK10® , and two samples were within the high range
values according to manual LT® and within the lower range values for i-TRACK10® .

2.4.2. Correlations between i-TRACK10® and Lisa-Tracker® Assays for ADA Detection
and Measurements
Linear correlations between ADA values obtained with different assays were between
0.59 and 0.77. For drugs, the differences were most significant within the highest ranges of
aIFX and aADAL values, but remained acceptable (Figure 1e–h). Analysis of systematic
bias from Bland–Altman plots indicated that aIFX levels were 50.7% higher on average
when using the i-TRACK10® analyzer compared with the manual LT® assay (95% CI: −74%,
176.3%), with one result outside the 95% CI range (−82.3, 182.3; bias 50, Figure 2e). For
Int. J. Mol. Sci. 2022, 23, 9561 8 of 12

drug levels, the observed differences were lower when comparing values obtained from
i-TRACK10® and DS2® LT® analyzers. Analysis of systematic bias from the Bland–Altman
plots indicated that aIFX levels were on average 9.3% higher using i-TRACK10® compared
with DS2® LT® (95% CI: −61.8%, 80.5%), with one point outside the 95% CI range (−56.3,
73.7; bias 8.7, Figure 2f). The average aADAL levels were 67.8% higher using i-TRACK10®
compared with the manual LT® assay (95% CI: −77.3%, 212.8%), with two results lying
outside the 95% CI range (−140.6, 335.8; bias 97.6, Figure 2g). There were only eight points
of comparison for aADAL values between i-TRACK10® and DS2® LT® , where the observed
trend included an equivalence of results between the two analyzers with no point lying
outside the 95% CI range (−71.3, 85.6; bias 7.1, Figure 2h).

2.5. Sample to Sample Carryover


The assessment of sample carryover phenomena did not reveal any cross-contamination
risks with the i-TRACK10® analyzer, because the carryover index calculated was always
less than 1% between runs (the obtained values were 0.28%, 0.59%, 0.03%, and 0.26% for
IFX, ADA, aIFX, and aADAL, respectively).

3. Discussion
This bi-centric study aimed to validate a new chemiluminescent immunoassay random-
access analyzer i-TRACK10® (Theradiag) to monitor treatment with IFX and ADAL, for
detection and quantification of drug levels and ADAs. These two drugs were chosen
because these treatments are frequently prescribed for many indications, notably in gas-
troenterology and rheumatology, and request for their dosage is very common in many
labs, not only in university hospital laboratories. We consider that these results can be
extrapolated to other molecules, for example, vedolizumab and ustekinumab.
Intra-run and inter-run comparisons were performed between i-TRACK10® and stan-
dard manual or automated (DS2® ) Lisa-Tracker® assays (both from Theradiag), in addition
to carryover phenomenon assessment. We found acceptable intra- and inter-run CV values
for all the parameters tested, which were similar between the two labs. Our acceptance
criterion was 20% CV as proposed by FDA guidelines [8]. The obtained CVs were consistent
with our previous evaluations of manual and DS2® Lisa-Tracker® assays, which showed
CV values between 5% for the minimal intra-run and 15% for the maximal inter-run results.
As expected, the inter-run CVs were globally superior to those obtained in the intra-run
assays, but there was no substantial difference concerning the imprecision of drug and
ADA values and between the different ranges of the datasets. The previous i-TRACK10®
evaluation by the manufacturer showed an intra-run CV ranging from 1.6% to 8.1% (run
of ten points) for IFX assay, and from 1.7% to 11.8% for the aIFX assay. For the inter-run
assays (six independent runs), CV ranged from 2% to 6.7% for IFX and from 2.3 to 4.8% for
aIFX [10].
The present study revealed an acceptable correlation between i-TRACK10® and auto-
mated or manual LT® assays, especially for lower concentration ranges, which represent
the most interesting group of results because these can impact clinical decision-making.
As shown in previous studies [11–15], the correlations were better for drugs than for their
corresponding ADAs (R2 ranged from 0.85 to 0.92 and from 0.59 to 0.77, respectively). The
lowest correlation was observed between i-TRACK10® and manual LT® values for aADAL,
with an R2 value of 0.59. The results were often higher for i-TRACK10® (on average 67.8%
higher), with a Bland–Altman bias at 97.6. This overestimation encountered when using
the i-TRACK10® analyzer was not mentioned in the manufacturer’s internal evaluation
report and is probably linked to the expanded measurement range.
We next tested whether the type of assay used could change the clinical interpretation
and subsequent attitude, especially in patients with chronic inflammatory bowel diseases.
With this aim, we classified our results into different concentration ranges based on thera-
peutic windows defined in several previous studies [16–21]. We found that different assays
suggested a change of category for 4% of the IFX samples, 17% of the ADAL samples,
Int. J. Mol. Sci. 2022, 23, 9561 9 of 12

3% of the aIFX samples, and 11% of the aADAL samples. We based our interpretation
on the cut-off used in clinical practice to define responder versus non-responder patients,
although these cut-off values are better defined for drugs [16,17,20,21] than for ADAs.
The kappa coefficient was globally acceptable for all the comparisons tested, according to
the acceptance criterion of Landis et al. [9]. Nevertheless, the type of assay, in particular
i-TRACK10® versus manual LT® assay, was found in our study to induce a change of
interpretation for ADA values, namely with a trend towards overestimation of ADA levels
using the i-TRACK10® analyzer. It is worth noting that drug and ADA monitoring tests are
not fully interchangeable, especially during the longitudinal follow-up of a given patient,
as previously reported by Pérez et al. [22]. A common clinical recommendation is to use
the same assay format for long-term follow-up. However, the observed differences in our
study particularly concern values within ranges, thus they have less potential to affect
clinical decision-making.
The i-TRACK10® analyzer presents many advantages, mentioned in Table 4, particu-
larly its expanded measurement ranges for ADAs with the highest point of the standard
curve approximately 10-fold higher than in the LT® kit. Another major advantage is the
rapidity of access to results. ELISA techniques require significant and incompressible
hands-on time and work by series, which significantly lengthens the time-to-result perfor-
mance parameter. The i-TRACK10® instrument has also benefits linked to standardized
techniques, with very low imprecision and personal or instrumental errors.

Table 4. List of the advantages and disadvantages of the manual and automatic Lisa-Tracker® and
I-TRACK10 systems.

DS2 L Tracker®
Manual Lisa Tracker® I-TRACK10®
(Specific to the Automated Process)
Random-access instrument:
decreased time to access the result
for clinicians (about 35 min to
obtain the first result)
Standardization thanks to the
Reliable and robust test to automated process
quantify drugs and anti-drug Reducing the chance of data input
antibodies errors with automated transmission
Precision, sensibility and No risk of human error affecting to the informatics lab system
Advantages
specificity of the dosage sample dilution and distribution Expansion of the measuring range,
Many molecules and their especially for ADA, useful for
specific antibodies are therapeutic de-escalation
available Precision, sensibility and specificity
of the dosage
Possibility of dosing multiple drugs
and anti-drugs antibodies in the
same round
Cost
Need to work in series
Unavoidable risk of technical
issues (manual dilution of
Risk of machine failure
samples, distribution, results
Delay of the accessibility to the
entry)
Disadvantages Risk of machine failure dosage in the instrument (for
Incompressible technical time
molecules infrequently
linked to manual steps
prescribed)
Increased time-to-result
(about 120 min to obtain the
first result)

Our study has some limitations, including the small number of samples tested, in
particular in the aADAL group compared between DS2® LT® and i-TRACK10® , thereby
Int. J. Mol. Sci. 2022, 23, 9561 10 of 12

limiting its overall strength somewhat. However, this explorative bi-centric study is the
first to have been devoted to evaluating the i-TRACK10® analyzer as a part of its future
accreditation process.

4. Methods
4.1. Patients and Samples
Samples were obtained from patients treated with IFX and ADAL for whom drug
levels and ADA concentrations were assessed as part of regular medical care, in two
different laboratories in France, CHU Saint-Etienne (laboratory A) and Bicêtre Hospital
at Le Kremlin-Bicêtre (laboratory B). Blood samples were collected immediately before
TNFi administration. These were centrifuged, and the patients’ sera were stored at −20 ◦ C
and thawed only once before use. The remaining samples from the routine laboratory
were selected and de-identified before use. Samples were used for the comparison assays
(different samples from the two labs), and the intra-run assays for laboratory A. Quality
controls (IMMUNO-TROL® , Theradiag, Croissy Beaubourg, France) were used for the
intra-run assays in laboratory B, and for the inter-run assays in both labs. The quality
controls (QC) were analyzed in the same manner as the patient samples, and were used at
two concentrations: a low/medium level (<4 µg/mL for drugs and <100 ng/mL for ADAs)
and a high level (>9 µg/mL for drugs and >100 ng/mL for ADAs). The number of samples
used for all evaluations is presented in Supplementary Table S1. No ethical approval was
required for the study, which was conducted for quality assurance purposes only (Code de
Santé Publique Français article L1243-3 and L1121-1).

4.2. Methods
Three assays were compared for this study: Lisa-Tracker® (LT) (Theradiag) with a
manual ELISA procedure (manual LT) (laboratory A); or with an automated ELISA DS2®
analyzer (DS2 LT) (Dynex technologies, Chantilly, VA, USA) (laboratory B); and the i-
TRACK10® analyzer (laboratories A and B). The LT procedure was performed according
to the manufacturer’s instructions and the protocol for the DS2 analyzer was identical to
the manual one. The distribution and washing steps were automated for the DS2, and the
optical density (OD) reading was performed using the spectrophotometer incorporated
inside the analyzer.

4.3. Data Analysis


As recommended by the quality assurance guidelines, the intermediate intra-run and
inter-run measurements of imprecision (CV, coefficient of variation (%)) of the i-TRACK10®
were evaluated for each of the parameters tested (IFX, ADAL, aIFX, aADAL). Intra-run and
inter-run variances for each parameter in the same lab were compared using Welch’s t-test,
and p < 0.05 was set as the level of significance.
For qualitative analysis, the agreement between results obtained with the i-TRACK10®
and the LT® technology was assessed using Cohen’s kappa coefficient, considering the
value of (a) as zero if there was no more agreement than could be expected by chance, or
(b) as 1 if there was a perfect agreement.
Kappa results were interpreted as follows: values lower than 0.2 indicated a slight
agreement, between 0.2 and 0.4 the agreement was considered as fair, between 0.4 and 0.6 as
moderate, between 0.6 and 0.8 as substantial, and values greater than 0.8 indicated almost
perfect agreement [9]. For quantitative comparison, linear regression was evaluated using
an x–y plot, and the R2 correlation coefficient was calculated. A value of 1 indicated perfect
linear correlation, while a value of 0 translated to an absence of correlation. Bland–Altman
plots were performed with GraphPad Prism version 8 (GraphPad Software, La Jolla, CA,
USA), and the mean difference between i-TRACK10® and other systems and the 95% limits
of agreement were calculated. A p-Value < 0.05 was considered statistically significant.
Sample carryover effect was assessed, according to the general quality assurance
protocol, by selection and analysis of two patient samples, one with high (H) and another
Int. J. Mol. Sci. 2022, 23, 9561 11 of 12

with low (L) values of IFX, ADAL, aADAL, and aIFX. The same H sample was analyzed
in triplicate followed by three analyses of the same L sample. Carryover was calculated
for each analyte as follows: ((L1-L3)/(H3-L3)) × 100. A value of carryover below 1% was
considered insignificant.

5. Conclusions
We performed a complete evaluation of a new random-access analyzer i-TRACK10®
for the monitoring of TNFi biologics. Despite good overall performances and correlations
between i-TRACK10® and Lisa-Tracker assays® , we recommend using the same assay
format for long-term follow-up of patients treated by TNFi biologics.

Supplementary Materials: The following supporting information can be downloaded at: https:
//www.mdpi.com/article/10.3390/ijms23179561/s1.
Author Contributions: Conceptualization, A.E.B., A.G., A.S. and S.P.; methodology, A.E.B., A.G.,
R.K. and S.H.-B.-A.; validation, A.E.B. and A.G.; formal analysis, A.E.B. and A.G.; investigation, L.W.,
R.K., X.R. and S.P.; writing—original draft preparation, A.E.B., A.G., A.S., R.K., S.H.-B.-A., X.R. and
S.P.; writing—review and editing, A.E.B., A.G., A.S., R.K., S.H.-B.-A., X.R. and S.P.; supervision, A.E.B.
and S.P. All authors have read and agreed to the published version of the manuscript.
Funding: This research received no external funding.
Informed Consent Statement: Informed consent was obtained from all subjects involved in the study.
Data Availability Statement: Datas are available upon request.
Acknowledgments: The authors thank Virginie Garnier for her technical support in this study.
Conflicts of Interest: The authors declare no conflict of interest to disclose.

Abbreviations
ADAL: adalimumab; aADAL: anti-adalimumab antibodies; CLIA: ChemiLuminescence ImmunoAssay;
CV: coefficient of variation; IFX: infliximab; aIFX: anti-infliximab antibodies; LT: Lisa-Tracker® ELISA
kit; LLOQ: Low Limit of Quantification; TNF: Tumor Necrosis Factor, TNFi: TNF inhibitors.

References
1. Lis, K.; Kuzawińska, O.; Bałkowiec-Iskra, E. Tumor necrosis factor inhibitors—State of knowledge. Arch. Med. Sci. AMS 2014, 10,
1175–1185. [CrossRef] [PubMed]
2. Udalova, I.; Monaco, C.; Nanchahal, J.; Feldmann, M. Anti-TNF Therapy. Microbiol. Spectr. 2016, 4, 4.4.46. [CrossRef]
3. Atiqi, S.; Hooijberg, F.; Loeff, F.C.; Rispens, T.; Wolbink, G.J. Immunogenicity of TNF-Inhibitors. Front. Immunol. 2020, 11, 312.
[CrossRef] [PubMed]
4. Vaisman-Mentesh, A.; Gutierrez-Gonzalez, M.; DeKosky, B.J.; Wine, Y. The Molecular Mechanisms That Underlie the Immune
Biology of Anti-drug Antibody Formation Following Treatment With Monoclonal Antibodies. Front. Immunol. 2020, 11, 1951.
[CrossRef] [PubMed]
5. Steenholdt, C.; Brynskov, J.; Thomsen, O.Ø.; Munck, L.K.; Fallingborg, J.; Christensen, L.A.; Pedersen, G.; Kjeldsen, J.; Jacobsen,
B.A.; Oxholm, A.S.; et al. Individualised therapy is more cost-effective than dose intensification in patients with Crohn’s disease
who lose response to anti-TNF treatment: A randomised, controlled trial. Gut 2014, 63, 919–927. [CrossRef] [PubMed]
6. Martelli, L.; Olivera, P.; Roblin, X.; Attar, A.; Peyrin-Biroulet, L. Cost-effectiveness of drug monitoring of anti-TNF therapy in
inflammatory bowel disease and rheumatoid arthritis: A systematic review. J. Gastroenterol. 2017, 52, 19–25. [CrossRef]
7. Gorovits, B.; Baltrukonis, D.J.; Bhattacharya, I.; Birchler, M.A.; Finco, D.; Sikkema, D.; Vincent, M.S.; Lula, S.; Marshall, L.;
Hickling, T.P. Immunoassay methods used in clinical studies for the detection of anti-drug antibodies to adalimumab and
infliximab. Clin. Exp. Immunol. 2018, 192, 348–365. [CrossRef]
8. Landis, J.R.; Koch, G.G. The measurement of observer agreement for categorical data. Biometrics 1977, 33, 159–174. [CrossRef]
9. Kadian, N.; Raju KS, R.; Rashid, M.; Malik, M.Y.; Taneja, I.; Wahajuddin, M. Comparative assessment of bioanalytical method
validation guidelines for pharmaceutical industry. J. Pharm. Biomed. Anal. 2016, 126, 83–97. [CrossRef]
10. Noguier, G.; Montaillier, C.; Daviere, S.; Colombeau, L.; Parussini, E. Development and evaluation of i-Tracker Infliximab and
i-Tracker Anti-Infliximab kits: Fast and innovative chemiluminescent assays for the monitoring of patients treated with Infliximab.
J. Crohns & Colitis 2020, 14, S563.
Int. J. Mol. Sci. 2022, 23, 9561 12 of 12

11. Ruiz-Argüello, B.; del Agua, A.R.; Torres, N.; Monasterio, A.; Martínez, A.; Nagore, D. Comparison study of two commercially
available methods for the determination of infliximab, adalimumab, etanercept and anti-drug antibody levels. Clin. Chem. Lab.
Med. CCLM 2013, 51, e287–e289. [CrossRef] [PubMed]
12. Bertin, D.; Serrero, M.; Grimaud, J.C.; Desjeux, A.; Desplat-Jégo, S. Monitoring of infliximab trough levels and anti-infliximab
antibodies in inflammatory bowel diseases: A comparison of three commercially available ELISA kits. Cytokine 2020, 126, 154859.
[CrossRef] [PubMed]
13. Laserna-Mendieta, E.J.; Salvador-Martín, S.; Marín-Jiménez, I.; Menchén, L.A.; López-Cauce, B.; López-Fernández, L.A.; Lucendo,
A.J. Comparison of a new rapid method for determination of serum anti-adalimumab and anti-infliximab antibodies with two
established ELISA kits. J. Pharm. Biomed. Anal. 2021, 198, 114003. [CrossRef] [PubMed]
14. Nasser, Y.; Labetoulle, R.; Harzallah, I.; Berger, A.E.; Roblin, X.; Paul, S. Comparison of Point-of-Care and Classical Immunoassays
for the Monitoring Infliximab and Antibodies Against Infliximab in IBD. Dig. Dis. Sci. 2018, 63, 2714–2721. [CrossRef]
15. Steenholdt, C.; Ainsworth, M.A.; Tovey, M.; Klausen, T.W.; Thomsen, O.Ø.; Brynskov, J.; Bendtzen, K. Comparison of techniques
for monitoring infliximab and antibodies against infliximab in Crohn’s disease. Ther. Drug Monit. 2013, 35, 530–538. [CrossRef]
16. Nakase, H.; Motoya, S.; Matsumoto, T.; Watanabe, K.; Hisamatsu, T.; Yoshimura, N.; Ishida, T.; Kato, S.; Nakagawa, T.; Esaki, M.;
et al. Significance of measurement of serum trough level and anti-drug antibody of adalimumab as personalised pharmacokinetics
in patients with Crohn’s disease: A subanalysis of the DIAMOND trial. Aliment. Pharmacol. Ther. 2017, 46, 873–882. [CrossRef]
17. Roblin, X.; Marotte, H.; Rinaudo, M.; Del Tedesco, E.; Moreau, A.; Phelip, J.M.; Genin, C.; Peyrin–Biroulet, L.; Paul, S. Association
between pharmacokinetics of adalimumab and mucosal healing in patients with inflammatory bowel diseases. Clin. Gastroenterol.
Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2014, 12, 80–84.e2. [CrossRef]
18. Pouw, M.F.; Krieckaert, C.L.; Nurmohamed, M.T.; van der Kleij, D.; Aarden, L.; Rispens, T.; Wolbink, G. Key findings towards
optimising adalimumab treatment: The concentration-effect curve. Ann. Rheum. Dis. 2015, 74, 513–518. [CrossRef]
19. Bortlik, M.; Duricova, D.; Malickova, K.; Machkova, N.; Bouzkova, E.; Hrdlicka, L.; Komarek, A.; Lukas, M. Infliximab trough
levels may predict sustained response to infliximab in patients with Crohn’s disease. J. Crohns Colitis 2013, 7, 736–743. [CrossRef]
20. Moore, C.; Corbett, G.; Moss, A.C. Systematic Review and Meta-Analysis: Serum Infliximab Levels During Maintenance Therapy
and Outcomes in Inflammatory Bowel Disease. J. Crohns Colitis 2016, 10, 619–625. [CrossRef]
21. Paul, S.; Del Tedesco, E.; Marotte, H.; Rinaudo-Gaujous, M.; Moreau, A.; Phelip, J.M.; Genin, C.; Peyrin-Biroulet, L.; Roblin, X.
Therapeutic drug monitoring of infliximab and mucosal healing in inflammatory bowel disease: A prospective study. Inflamm.
Bowel Dis. 2013, 19, 2568–2576. [CrossRef] [PubMed]
22. Pérez, I.; Fernández, L.; Sánchez-Ramón, S.; Alba, C.; Zatarain, A.; Cañas, M.; López, O.N.; Olivares, D.; Rey, E.; Taxonera, C.
Reliability evaluation of four different assays for therapeutic drug monitoring of infliximab levels. Ther. Adv. Gastroenterol. 2018,
11, 1756284818783613. [CrossRef] [PubMed]

You might also like